1
|
Zhang Z, Li L, Shi H, Chen B, Li X, Zhang Y, Liu F, Wei W, Zhou Y, Liu K, Xia W, Gu X, Huang J, Tu S, Yin C, Shao A, Jiang L. Role of Circular RNAs in Atherosclerosis through Regulation of Inflammation, Cell Proliferation, Migration, and Apoptosis: Focus on Atherosclerotic Cerebrovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1461. [PMID: 37629751 PMCID: PMC10456328 DOI: 10.3390/medicina59081461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/29/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023]
Abstract
Atherosclerosis (AS) is a disease dangerous to human health and the main pathological cause of ischemic cardiovascular diseases. Although its pathogenesis is not fully understood, numerous basic and clinical studies have shown that AS is a chronic inflammatory disease existing in all stages of atherogenesis. It may be a common link or pathway in the pathogenesis of multiple atherogenic factors. Inflammation is associated with AS complications, such as plaque rupture and ischemic cerebral infarction. In addition to inflammation, apoptosis plays an important role in AS. Apoptosis is a type of programmed cell death, and different apoptotic cells have different or even opposite roles in the process of AS. Unlike linear RNA, circular RNA (circRNA) a covalently closed circular non-coding RNA, is stable and can sponge miRNA, which can affect the stages of AS by regulating downstream pathways. Ultimately, circRNAs play very important roles in AS by regulating inflammation, apoptosis, and some other mechanisms. The study of circular RNAs can provide new ideas for the prediction, prevention, and treatment of AS.
Collapse
Affiliation(s)
- Zheng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Lingfei Li
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Huanqing Shi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Biao Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Xiaoqin Li
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Yuyao Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Fei Liu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Wan Wei
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Yongji Zhou
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Keqin Liu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Wenqing Xia
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Xin Gu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Jinyu Huang
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China;
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310006, China;
| | - Congguo Yin
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Disease, Hangzhou 310009, China
| | - Lin Jiang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| |
Collapse
|
2
|
Tian JJ, Levy M, Zhang X, Sinnott R, Maddela R. Counteracting Health Risks by Modulating Homeostatic Signaling. Pharmacol Res 2022; 182:106281. [PMID: 35661711 DOI: 10.1016/j.phrs.2022.106281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/14/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
Homeostasis was initially conceptualized by Bernard and Cannon around a century ago as a steady state of physiological parameters that vary within a certain range, such as blood pH, body temperature, and heart rate1,2. The underlying mechanisms that maintain homeostasis are explained by negative feedbacks that are executed by the neuronal, endocrine, and immune systems. At the cellular level, homeostasis, such as that of redox and energy steady state, also exists and is regulated by various cell signaling pathways. The induction of homeostatic mechanism is critical for human to adapt to various disruptive insults (stressors); while on the other hand, adaptation occurs at the expense of other physiological processes and thus runs the risk of collateral damages, particularly under conditions of chronic stress. Conceivably, anti-stress protection can be achieved by stressor-mimicking medicinals that elicit adaptive responses prior to an insult and thereby serve as health risk countermeasures; and in situations where maladaptation may occur, downregulating medicinals could be used to suppress the responses and prevent subsequent pathogenesis. Both strategies are preemptive interventions particularly suited for individuals who carry certain lifestyle, environmental, or genetic risk factors. In this article, we will define and characterize a new modality of prophylactic intervention that forestalls diseases via modulating homeostatic signaling. Moreover, we will provide evidence from the literature that support this concept and distinguish it from other homeostasis-related interventions such as adaptogen, hormesis, and xenohormesis.
Collapse
Affiliation(s)
- Junqiang J Tian
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA.
| | - Mark Levy
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| | - Xuekai Zhang
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing100029, China; US Center for Chinese Medicine, 14801 Physicians lane, 171 A 2nd Floor, #281, Rockville MD 20850, USA
| | - Robert Sinnott
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| | - Rolando Maddela
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| |
Collapse
|
3
|
Chronic obstructive pulmonary disease and atherosclerosis: common mechanisms and novel therapeutics. Clin Sci (Lond) 2022; 136:405-423. [PMID: 35319068 PMCID: PMC8968302 DOI: 10.1042/cs20210835] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/17/2022] [Accepted: 03/07/2022] [Indexed: 12/17/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and atherosclerosis are chronic irreversible diseases, that share a number of common causative factors including cigarette smoking. Atherosclerosis drastically impairs blood flow and oxygen availability to tissues, leading to life-threatening outcomes including myocardial infarction (MI) and stroke. Patients with COPD are most likely to die as a result of a cardiovascular event, with 30% of all COPD-related deaths being attributed to cardiovascular disease (CVD). Both atherosclerosis and COPD involve significant local (i.e. lung, vasculature) and systemic inflammation and oxidative stress, of which current pharmacological treatments have limited efficacy, hence the urgency for the development of novel life-saving therapeutics. Currently these diseases must be treated individually, with no therapies available that can effectively reduce the likelihood of comorbid CVD other than cessation of cigarette smoking. In this review, the important mechanisms that drive atherosclerosis and CVD in people with COPD are explained and we propose that modulation of both the oxidative stress and the inflammatory burden will provide a novel therapeutic strategy to treat both the pulmonary and systemic manifestations related to these diseases.
Collapse
|
4
|
Ma J, Zhang Y, Sugai T, Kubota T, Keino H, El-Salhy M, Ozaki M, Umezawa K. Inhibition of Cellular and Animal Inflammatory Disease Models by NF-κB Inhibitor DHMEQ. Cells 2021; 10:2271. [PMID: 34571920 PMCID: PMC8466912 DOI: 10.3390/cells10092271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
General inflammatory diseases include skin inflammation, rheumatoid arthritis, inflammatory bowel diseases, sepsis, arteriosclerosis, and asthma. Although these diseases have been extensively studied, most of them are still difficult to treat. Meanwhile, NF-κB is a transcription factor promoting the expression of many inflammatory mediators. NF-κB is likely to be involved in the mechanism of most inflammatory diseases. We discovered a specific NF-κB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), about 20 years ago by molecular design from a natural product. It directly binds to and inactivates NF-κB components. It has been widely used to suppress cellular and animal inflammatory disease models and was shown to be potent in vivo anti-inflammatory activity without any toxicity. We have prepared ointment of DHMEQ for the treatment of severe skin inflammation. It inhibited inflammatory cytokine expressions and lowered the clinical score in mouse models of atopic dermatitis. Intraperitoneal (IP) administration of DHMEQ ameliorated various disease models of inflammation, such as rheumatoid arthritis, sepsis, and also graft rejection. It has been suggested that inflammatory cells in the peritoneal cavity would be important for most peripheral inflammation. In the present review, we describe the synthesis, mechanism of action, and cellular and in vivo anti-inflammatory activities and discuss the clinical use of DHMEQ for inflammatory diseases.
Collapse
Affiliation(s)
- Jun Ma
- Shenzhen Wanhe Pharmaceutical Co., Ltd., Shenzhen 518107, China;
| | - Yuyang Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China;
| | - Takeshi Sugai
- Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan;
| | - Tetsuo Kubota
- Department of Medical Technology, Tsukuba International University, Tsuchiura 300-0051, Japan;
| | - Hiroshi Keino
- Department of Ophthalmology, Kyorin University School of Medicine, Tokyo 181-8611, Japan;
| | - Magdy El-Salhy
- Department of Medicine, Stord Helse-Fonna Hospital, Tysevegen 64, 54 16 Stord, Norway;
| | - Michitaka Ozaki
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido University, Sapporo 060-0812, Japan;
| | - Kazuo Umezawa
- Department of Molecular Target Medicine, Aichi Medical University, Nagakute 480-1195, Japan
| |
Collapse
|
5
|
Blockade of NF-κB Translocation and of RANKL/RANK Interaction Decreases the Frequency of Th2 and Th17 Cells Capable of IL-4 and IL-17 Production, Respectively, in a Mouse Model of Allergic Asthma. Molecules 2021; 26:molecules26113117. [PMID: 34071080 PMCID: PMC8197166 DOI: 10.3390/molecules26113117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 11/23/2022] Open
Abstract
The main purpose of this study was to investigate whether the blockade of the interaction between the receptor activator of nuclear factor-κB (NF-ĸB) ligand (RANKL) and its receptor RANK as well as the blockade of NF-κB inhibitor kinase (IKK) and of NF-κB translocation have the potential to suppress the pathogenesis of allergic asthma by inhibition and/or enhancement of the production by CD4+ and CD8+ T cells of important cytokines promoting (i.e., IL-4 and IL-17) and/or inhibiting (i.e., IL-10 and TGF-β), respectively, the development of allergic asthma. Studies using ovalbumin(OVA)-immunized mice have demonstrated that all the tested therapeutic strategies prevented the OVA-induced increase in the absolute number of IL-4- and IL-17-producing CD4+ T cells (i.e., Th2 and Th17 cells, respectively) indirectly, i.e., through the inhibition of the clonal expansion of these cells in the mediastinal lymph nodes. Additionally, the blockade of NF-κB translocation and RANKL/RANK interaction, but not IKK, prevented the OVA-induced increase in the percentage of IL-4-, IL-10- and IL-17-producing CD4+ T cells. These latter results strongly suggest that both therapeutic strategies can directly decrease IL-4 and IL-17 production by Th2 and Th17 cells, respectively. This action may constitute an important mechanism underlying the anti-asthmatic effect induced by the blockade of NF-κB translocation and of RANKL/RANK interaction. Thus, in this context, both these therapeutic strategies seem to have an advantage over the blockade of IKK. None of the tested therapeutic strategies increased both the absolute number and frequency of IL-10- and TGF-β-producing Treg cells, and hence they lacked the potential to inhibit the development of the disease via this mechanism.
Collapse
|
6
|
Gregorczyk I, Maślanka T. Blockade of RANKL/RANK and NF-ĸB signalling pathways as novel therapeutic strategies for allergic asthma: A comparative study in a mouse model of allergic airway inflammation. Eur J Pharmacol 2020; 879:173129. [PMID: 32339516 DOI: 10.1016/j.ejphar.2020.173129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022]
Abstract
The main aims of this study were: (1) to investigate whether a blockade of the interaction between the receptor activator of nuclear factor-κB (NF-ĸB) ligand (RANKL) and its receptor RANK may have potential as a novel therapeutic strategy for allergic asthma; (2) to compare the efficacies of the blockade of RANKL/RANK interaction as well as the blockade of NF-κB inhibitor kinase (IKK) and of NF-κB translocation to the nucleus, also in comparison with glucocorticosteroid treatment, in terms of the development of a mouse model of allergic airway inflammation (AAI) and accompanying immune response. The blockade of each of the targets fully prevented the development of AAI. All the tested therapeutic strategies seemed to have a certain advantage over glucocorticosteroids with regard to counteracting the development of AAI. Prevention of the activation and clonal expansion of CD4+ effector T (Teff) cells in the mediastinal lymph nodes (MLNs) constitutes a fundamental event underlying the anti-asthmatic effect induced by the blockade of IKK, NF-κB translocation or of RANKL/RANK interaction. The results indicate that attenuation of the CD11b+CD103-CD11chigh dendritic cell response in the MLNs is an initial but not the main mechanism responsible for this effect. In turn, the direct anti-proliferative action on CD4+ Teff cells seems to constitute the chief mechanism responsible for the anti-asthmatic effect of all the tested therapeutic strategies. A clinical implication is that local inhibition of RANKL/RANK interaction achieved via inhalatory administration of a RANKL antagonist can be considered as a novel therapeutic strategy in treatment of allergic asthma.
Collapse
Affiliation(s)
- Izabela Gregorczyk
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Street 13, 10-719, Olsztyn, Poland
| | - Tomasz Maślanka
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Street 13, 10-719, Olsztyn, Poland.
| |
Collapse
|
7
|
Ando Y, Keino H, Kudo A, Hirakata A, Okada AA, Umezawa K. Anti-Inflammatory Effect of Dehydroxymethylepoxyquinomicin, a Nuclear factor-κB Inhibitor, on Endotoxin-Induced Uveitis in Rats In vivo and In vitro. Ocul Immunol Inflamm 2019; 28:240-248. [PMID: 30950670 DOI: 10.1080/09273948.2019.1568502] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Purpose: To determine the anti-inflammatory effects of dehydroxymethylepoxyquinomicin (DHMEQ), a nuclear factor-κB (NF-κB) inhibitor, on endotoxin-induced uveitis (EIU) in rats.Methods: EIU was induced by a subcutaneous injection of lipopolysaccharide (LPS) in Lewis rats. DHMEQ was injected intraperitoneally concurrently with the LPS. Aqueous humor was collected 24 h after the LPS injection. Isolated peritoneal exudate cells (PECs) were exposed to LPS with or without DHMEQ to determine the production of TNF-α, IL-6, and MCP-1.Results: DHMEQ significantly reduced the number of infiltrating cells, and the concentrations of proteins, TNF-α, and IL-6 in the aqueous humor. DHMEQ suppressed the production of TNF-α, IL-6, and MCP-1 from PECs. Immunochemistry revealed a reduction in the translocation of the NF-κB p65 into the nuclei in DHMEQ-exposed PECs.Conclusions: The results indicate that DHMEQ has anti-inflammatory effects on EIU and may be a promising agent to treat intraocular inflammation.
Collapse
Affiliation(s)
- Yoshimasa Ando
- Department of Ophthalmology, Kyorin University School of Medicine, Tokyo, Japan
| | - Hiroshi Keino
- Department of Ophthalmology, Kyorin University School of Medicine, Tokyo, Japan
| | - Akihiko Kudo
- Department of Anatomy, Kyorin University School of Medicine, Tokyo, Japan
| | - Akito Hirakata
- Department of Ophthalmology, Kyorin University School of Medicine, Tokyo, Japan
| | - Annabelle A Okada
- Department of Ophthalmology, Kyorin University School of Medicine, Tokyo, Japan
| | - Kazuo Umezawa
- Department of Molecular Target Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
8
|
Wang P, Liu Z, Zhang X, Li J, Sun L, Ju Z, Li J, Chan P, Liu GH, Zhang W, Song M, Qu J. CRISPR/Cas9-mediated gene knockout reveals a guardian role of NF-κB/RelA in maintaining the homeostasis of human vascular cells. Protein Cell 2018; 9:945-965. [PMID: 29968158 PMCID: PMC6208479 DOI: 10.1007/s13238-018-0560-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 06/08/2018] [Indexed: 12/12/2022] Open
Abstract
Vascular cell functionality is critical to blood vessel homeostasis. Constitutive NF-κB activation in vascular cells results in chronic vascular inflammation, leading to various cardiovascular diseases. However, how NF-κB regulates human blood vessel homeostasis remains largely elusive. Here, using CRISPR/Cas9-mediated gene editing, we generated RelA knockout human embryonic stem cells (hESCs) and differentiated them into various vascular cell derivatives to study how NF-κB modulates human vascular cells under basal and inflammatory conditions. Multi-dimensional phenotypic assessments and transcriptomic analyses revealed that RelA deficiency affected vascular cells via modulating inflammation, survival, vasculogenesis, cell differentiation and extracellular matrix organization in a cell type-specific manner under basal condition, and that RelA protected vascular cells against apoptosis and modulated vascular inflammatory response upon tumor necrosis factor α (TNFα) stimulation. Lastly, further evaluation of gene expression patterns in IκBα knockout vascular cells demonstrated that IκBα acted largely independent of RelA signaling. Taken together, our data reveal a protective role of NF-κB/RelA in modulating human blood vessel homeostasis and map the human vascular transcriptomic landscapes for the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Ping Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoqian Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingyi Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Liang Sun
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Jian Li
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Piu Chan
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Guang-Hui Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
- Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| | - Weiqi Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
9
|
Li B, Li W, Li X, Zhou H. Inflammation: A Novel Therapeutic Target/Direction in Atherosclerosis. Curr Pharm Des 2018; 23:1216-1227. [PMID: 28034355 PMCID: PMC6302344 DOI: 10.2174/1381612822666161230142931] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/27/2016] [Indexed: 12/27/2022]
Abstract
Over the past two decades, the viewpoint of atherosclerosis has been replaced gradually by a lipid-driven, chronic, low-grade inflammatory disease of the arterial wall. Current treatment of atherosclerosis is focused on limiting its risk factors, such as hyperlipidemia or hypertension. However, treatment targeting the inflammatory nature of atherosclerosis is still very limited and deserves further attention to fight atherosclerosis successfully. Here, we review the current development of inflammation and atherosclerosis to discuss novel insights and potential targets in atherosclerosis, and to address drug discovery based on anti-inflammatory strategy in atherosclerotic disease.
Collapse
Affiliation(s)
- Bin Li
- Department of Pharmacology, College of Pharmacy, Third Military Medical University, Chongqing 400038. China
| | - Weihong Li
- Assisted Reproductive Center, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016. China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Third Military Medical University, Chongqing 400038. China
| | - Hong Zhou
- Department of Pharmacology, College of Pharamacy, The Third Military Medical University, P.O. Box: 400038, Chongqing. China
| |
Collapse
|
10
|
Effect of Kangshuanyihao Formula on the Inflammatory Reaction and SIRT1/TLR4/NF- κB Signaling Pathway in Endothelial Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:9019765. [PMID: 28539968 PMCID: PMC5429935 DOI: 10.1155/2017/9019765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/20/2022]
Abstract
Endothelial injury plays an important role in atherosclerosis (AS). Kangshuanyihao formula uses therapeutic principles from Chinese medicine to supplement Qi, thereby promoting blood circulation, and remove blood stasis. The mechanism by which the formula inhibits endothelial injury was examined in a rat model of 1,25-dihydroxyvitamin D3 (VD3) intraperitoneal injection and high-fat-induced endothelial injury. Rats were randomly divided into the model, high-dose, middle-dose, low-dose, positive drug (rosuvastatin), and combination (positive drug + middle-dose) groups; 10 Sprague-Dawley rats served as the blank group. The aortic endothelium was stained with hematoxylin and eosin and the levels of blood lipids and inflammation markers (mRNA and protein) were measured. Endothelial injury, lipid levels, and inflammation were increased in the model. Kangshuanyihao formula reduced endothelial injury, improved lipid levels, and downregulated inflammation, as shown by significant reduction of the protein levels of SIRT1, TLR4, and NF-κB and mRNA levels of SIRT1, TLR4, NF-κB, IL-1β, IL-6, and IL-12. Thus, we conclude that Kangshuanyihao formula can inhibit the inflammatory reaction in the rat model of high-fat-induced endothelial injury after intraperitoneal injection of VD3. This mechanism may be attributed to regulating the SIRT1/TLR4/NF-κB signaling pathway.
Collapse
|
11
|
Yamanouchi S, Adachi Y, Shimo T, Umezawa K, Okigaki M, Tsuji S, Li M, Takaya J, Kuge T, Ikehara S, Kaneko K. A nuclear factor-κB inhibitor, dehydroxymethylepoxyquinomicin, ameliorates GVHD in allogeneic bone marrow transplantation. Immunobiology 2015; 220:1059-1066. [PMID: 26004346 DOI: 10.1016/j.imbio.2015.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 04/11/2015] [Accepted: 05/01/2015] [Indexed: 10/23/2022]
Abstract
GVHD is a crucial mortality factor in allogeneic bone marrow transplantation (ABMT). In this paper, we show that dehydroxymethylepoxyquinomicin (DHMEQ), a novel inhibitor of nuclear factor-κB, suppresses GVHD, resulting in an improved mortality rate in a mouse ABMT model. Bone marrow cells from C57BL/6 mice (B6 mice) were transplanted into lethally irradiated BALB/c mice. Two weeks later, spleen cells from B6 mice were transplanted into the irradiated BALB/c mice. From one week after the injection of spleen cells, when the mice started to show GVHD, the mice were also injected intraperitoneally daily with DHMEQ or vehicle only (DMSO) for 4 weeks. By 80 days after the ABMT, 6/14 of the vehicle-injected mice (43%) had died because of GVHD, whereas all DHMEQ-injected mice survived this observation period and developed milder GVHD than the vehicle-injected mice. When regulatory T cells were reduced by the injection of anti-folate receptor 4 (FR4) antibody, the effects of DHMEQ were reduced. These findings suggest that administration of DHMEQ could become a new strategy for preventing fatalities from GVHD.
Collapse
Affiliation(s)
- Sohsaku Yamanouchi
- Department of Pediatrics, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| | - Yasushi Adachi
- Division of Surgical Pathology, Toyooka Hospital, Toyooka City, Hyogo 668-8501, Japan; Department of Stem Cell Disorders, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan.
| | - Tomohiko Shimo
- Department of Pediatrics, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| | - Kazuo Umezawa
- Department of Molecular Target Medicine Screening, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Mitsuhiko Okigaki
- Department of Internal Medicine, Otokoyama Hospital, Yawata City, Kyoto 614-8366, Japan
| | - Shoji Tsuji
- Department of Pediatrics, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| | - Ming Li
- Department of Stem Cell Disorders, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| | - Junji Takaya
- Department of Pediatrics, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| | - Tomohiro Kuge
- Department of Clinical Laboratory, Toyooka Hospital, Tobera, Toyooka City, Hyogo 668-8501, Japan
| | - Susumu Ikehara
- Department of Stem Cell Disorders, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| |
Collapse
|
12
|
Tilstam PV, Gijbels MJ, Habbeddine M, Cudejko C, Asare Y, Theelen W, Zhou B, Döring Y, Drechsler M, Pawig L, Simsekyilmaz S, Koenen RR, de Winther MPJ, Lawrence T, Bernhagen J, Zernecke A, Weber C, Noels H. Bone marrow-specific knock-in of a non-activatable Ikkα kinase mutant influences haematopoiesis but not atherosclerosis in Apoe-deficient mice. PLoS One 2014; 9:e87452. [PMID: 24498325 PMCID: PMC3911989 DOI: 10.1371/journal.pone.0087452] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/27/2013] [Indexed: 12/13/2022] Open
Abstract
Background The Ikkα kinase, a subunit of the NF-κB-activating IKK complex, has emerged as an important regulator of inflammatory gene expression. However, the role of Ikkα-mediated phosphorylation in haematopoiesis and atherogenesis remains unexplored. In this study, we investigated the effect of a bone marrow (BM)-specific activation-resistant Ikkα mutant knock-in on haematopoiesis and atherosclerosis in mice. Methods and Results Apolipoprotein E (Apoe)-deficient mice were transplanted with BM carrying an activation-resistant Ikkα gene (IkkαAA/AAApoe−/−) or with Ikkα+/+Apoe−/− BM as control and were fed a high-cholesterol diet for 8 or 13 weeks. Interestingly, haematopoietic profiling by flow cytometry revealed a significant decrease in B-cells, regulatory T-cells and effector memory T-cells in IkkαAA/AAApoe−/− BM-chimeras, whereas the naive T-cell population was increased. Surprisingly, no differences were observed in the size, stage or cellular composition of atherosclerotic lesions in the aorta and aortic root of IkkαAA/AAApoe−/− vs Ikkα+/+Apoe−/− BM-transplanted mice, as shown by histological and immunofluorescent stainings. Necrotic core sizes, apoptosis, and intracellular lipid deposits in aortic root lesions were unaltered. In vitro, BM-derived macrophages from IkkαAA/AAApoe−/− vs Ikkα+/+Apoe−/− mice did not show significant differences in the uptake of oxidized low-density lipoproteins (oxLDL), and, with the exception of Il-12, the secretion of inflammatory proteins in conditions of Tnf-α or oxLDL stimulation was not significantly altered. Furthermore, serum levels of inflammatory proteins as measured with a cytokine bead array were comparable. Conclusion Our data reveal an important and previously unrecognized role of haematopoietic Ikkα kinase activation in the homeostasis of B-cells and regulatory T-cells. However, transplantation of IkkαAA mutant BM did not affect atherosclerosis in Apoe−/− mice. This suggests that the diverse functions of Ikkα in haematopoietic cells may counterbalance each other or may not be strong enough to influence atherogenesis, and reveals that targeting haematopoietic Ikkα kinase activity alone does not represent a therapeutic approach.
Collapse
Affiliation(s)
- Pathricia V. Tilstam
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Marion J. Gijbels
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mohamed Habbeddine
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Marseille, France
| | - Céline Cudejko
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Marseille, France
| | - Yaw Asare
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Wendy Theelen
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Baixue Zhou
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maik Drechsler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lukas Pawig
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Sakine Simsekyilmaz
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Rory R. Koenen
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Menno P. J. de Winther
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Toby Lawrence
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, Marseille, France
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
- August-Lenz-Stiftung, Institute for Cardiovascular Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Alma Zernecke
- Rudolf Virchow Center and Institute of Clinical Biochemistry and Pathobiochemistry, University of Würzburg, Würzburg, Germany
- Department of Vascular Surgery, Klinikum rechts der Isar Technical University Munich, Munich, Germany
- German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
- * E-mail: (CW); (HN)
| | - Heidi Noels
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
- * E-mail: (CW); (HN)
| |
Collapse
|
13
|
Effect of low extracellular pH on NF-κB activation in macrophages. Atherosclerosis 2014; 233:537-544. [PMID: 24530961 PMCID: PMC3989994 DOI: 10.1016/j.atherosclerosis.2014.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 12/12/2013] [Accepted: 01/08/2014] [Indexed: 02/02/2023]
Abstract
Objective Many diseases, including atherosclerosis, involve chronic inflammation. The master transcription factor for inflammation is NF-κB. Inflammatory sites have a low extracellular pH. Our objective was to demonstrate the effect of pH on NF-κB activation and cytokine secretion. Methods Mouse J774 macrophages or human THP-1 or monocyte-derived macrophages were incubated at pH 7.0–7.4 and inflammatory cytokine secretion and NF-κB activity were measured. Results A pH of 7.0 greatly decreased pro-inflammatory cytokine secretion (TNF or IL-6) by J774 macrophages, but not THP-1 or human monocyte-derived macrophages. Upon stimulation of mouse macrophages, the levels of IκBα, which inhibits NF-κB, fell but low pH prevented its later increase, which normally restores the baseline activity of NF-κB, even though the levels of mRNA for IκBα were increased. pH 7.0 greatly increased and prolonged NF-κB binding to its consensus promoter sequence, especially the anti-inflammatory p50:p50 homodimers. Human p50 was overexpressed using adenovirus in THP-1 macrophages and monocyte-derived macrophages to see if it would confer pH sensitivity to NF-κB activity in human cells. Overexpression of p50 increased p50:p50 DNA-binding and in THP-1 macrophages inhibited considerably TNF and IL-6 secretion, but there was still no effect of pH on p50:p50 DNA binding or cytokine secretion. Conclusion A modest decrease in pH can sometimes have marked effects on NF-κB activation and cytokine secretion and might be one reason to explain why mice normally develop less atherosclerosis than do humans. Low extracellular pH decreased cytokine secretion by mouse macrophages. IκBα, which inhibits NF-κB, fell but low pH prevented its later increase. Low pH prolonged anti-inflammatory p50:p50 homodimer binding to an NF-κB promoter. Overexpression of p50 increased p50:p50 DNA-binding and inhibited TNF secretion. A modest decrease in pH can have marked effects on NF-κB activation.
Collapse
|
14
|
Morrison M, van der Heijden R, Heeringa P, Kaijzel E, Verschuren L, Blomhoff R, Kooistra T, Kleemann R. Epicatechin attenuates atherosclerosis and exerts anti-inflammatory effects on diet-induced human-CRP and NFκB in vivo. Atherosclerosis 2014; 233:149-56. [PMID: 24529136 DOI: 10.1016/j.atherosclerosis.2013.12.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 12/11/2013] [Accepted: 12/22/2013] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Previous studies investigating flavanol-rich foods provide indications for potential cardioprotective effects of these foods, but the effects of individual flavanols remain unclear. We investigated whether the flavanol epicatechin can reduce diet-induced atherosclerosis, with particular emphasis on the cardiovascular risk factors dyslipidaemia and inflammation. METHODS ApoE*3-Leiden mice were fed a cholesterol-containing atherogenic diet with or without epicatechin (0.1% w/w) to study effects on early- and late-stage atherosclerosis (8 w and 20 w). In vivo effects of epicatechin on diet-induced inflammation were studied in human-CRP transgenic mice and NFκB-luciferase reporter mice. RESULTS Epicatechin attenuated atherosclerotic lesion area in ApoE*3-Leiden mice by 27%, without affecting plasma lipids. This anti-atherogenic effect of epicatechin was specific to the severe lesion types, with no effect on mild lesions. Epicatechin mitigated diet-induced increases in plasma SAA (in ApoE*3-Leiden mice) and plasma human-CRP (in human-CRP transgenic mice). Microarray analysis of aortic gene expression revealed an attenuating effect of epicatechin on several diet-induced pro-atherogenic inflammatory processes in the aorta (e.g. chemotaxis of cells, matrix remodelling), regulated by NFκB. These findings were confirmed immunohistochemically by reduced lesional neutrophil content in HCE, and by inhibition of diet-induced NFκB activity in epicatechin-treated NFκB-luciferase reporter mice. CONCLUSION Epicatechin attenuates development of atherosclerosis and impairs lesion progression from mild to severe lesions in absence of an effect on dyslipidaemia. The observed reduction of circulating inflammatory risk factors by epicatechin (e.g. SAA, human-CRP), as well as its local anti-inflammatory activity in the vessel wall, provide a rationale for epicatechin's anti-atherogenic effects.
Collapse
Affiliation(s)
- Martine Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK Leiden, The Netherlands; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; Top Institute Food and Nutrition, Nieuwe Kanaal 9A, 6709 PA Wageningen, The Netherlands.
| | - Roel van der Heijden
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; Top Institute Food and Nutrition, Nieuwe Kanaal 9A, 6709 PA Wageningen, The Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands
| | - Eric Kaijzel
- Molecular Endocrinology and Molecular Imaging, Department of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Lars Verschuren
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK Leiden, The Netherlands
| | - Rune Blomhoff
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, 0372 Oslo, Norway
| | - Teake Kooistra
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK Leiden, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK Leiden, The Netherlands
| |
Collapse
|
15
|
Nohria A, Kinlay S, Buck JS, Redline W, Copeland-Halperin R, Kim S, Beckman JA. The effect of salsalate therapy on endothelial function in a broad range of subjects. J Am Heart Assoc 2014; 3:e000609. [PMID: 24390146 PMCID: PMC3959688 DOI: 10.1161/jaha.113.000609] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Inflammation is fundamental to the development of atherosclerosis. We examined the effect of anti‐inflammatory doses of salicylate on endothelium‐dependent vasodilation, a biomarker of cardiovascular risk, in a broad range of subjects. Methods and Results We performed a randomized, double‐blind, placebo‐controlled crossover trial evaluating the effects of 4 weeks of high‐dose salsalate (disalicylate) therapy on endothelium‐dependent flow‐mediated and endothelium‐independent vasodilation. Fifty‐eight subjects, including 17 with metabolic syndrome, 13 with atherosclerosis, and 28 healthy controls, were studied. Among all subjects, endothelium‐dependent flow‐mediated vasodilation decreased after salsalate compared with placebo therapy (P=0.01), whereas nitroglycerin‐mediated, endothelium‐independent vasodilation was unchanged (P=0.97). Endothelium‐dependent flow‐mediated vasodilation after salsalate therapy was impaired compared with placebo therapy in subjects with therapeutic salicylate levels (n=31, P<0.02) but not in subjects with subtherapeutic levels (P>0.2). Conclusions Salsalate therapy, particularly when therapeutic salicylate levels are achieved, impairs endothelium‐dependent vasodilation in a broad range of subjects. These data raise concern about the possible deleterious effects of anti‐inflammatory doses of salsalate on cardiovascular risk. Clinical Trial Registration URL: www.clinicaltrials.gov. Unique Identifiers: NCT00760019 and NCT00762827.
Collapse
Affiliation(s)
- Anju Nohria
- Cardiovascular Medicine, Department of Internal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | | | | | | |
Collapse
|
16
|
Shinozaki S, Chiba T, Kokame K, Miyata T, Kaneko E, Shimokado K. A deficiency of Herp, an endoplasmic reticulum stress protein, suppresses atherosclerosis in ApoE knockout mice by attenuating inflammatory responses. PLoS One 2013; 8:e75249. [PMID: 24204574 PMCID: PMC3810372 DOI: 10.1371/journal.pone.0075249] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 08/13/2013] [Indexed: 01/18/2023] Open
Abstract
Herp was originally identified as an endoplasmic reticulum (ER) stress protein in vascular endothelial cells. ER stress is induced in atherosclerotic lesions, but it is not known whether Herp plays any role in the development of atherosclerosis. To address this question, we generated Herp- and apolipoprotein E (apoE)-deficient mice (Herp(-/-); apoE(-/-) mice) by crossbreeding Herp(-/-) mice and apoE(-/-) mice. Herp was expressed in the endothelial cells and medial smooth muscle cells of the aorta, as well as in a subset of macrophages in the atherosclerotic lesions in apoE(-/-) mice, while there was no expression of Herp in the Herp(-/-); apoE(-/-) mice. The doubly deficient mice developed significantly fewer atherosclerotic lesions than the apoE(-/-) mice at 36 and 72 weeks of age, whereas the plasma levels of cholesterol and triglycerides were not significantly different between the strains. The plasma levels of non-esterified fatty acids were significantly lower in the Herp(-/-); apoE(-/-) mice when they were eight and 16 weeks old. The gene expression levels of ER stress response proteins (GRP78 and CHOP) and inflammatory cytokines (IL-1β, IL-6, TNF-α and MCP-1) in the aorta were significantly lower in Herp(-/-); apoE(-/-) mice than in apoE(-/-) mice, suggesting that Herp mediated ER stress-induced inflammation. In fact, peritoneal macrophages isolated from Herp-deficient mice and RAW264.7 macrophages in which Herp was eliminated with a siRNA expressed lower levels of mRNA for inflammatory cytokines when they were treated with tunicamycin. Herp deficiency affected the major mediators of the unfolded protein response, including IRE1 and PERK, but not ATF6. These findings suggest that a deficiency of Herp suppressed the development of atherosclerosis by attenuating the ER stress-induced inflammatory reactions.
Collapse
Affiliation(s)
- Shohei Shinozaki
- Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Tsuyoshi Chiba
- Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
- Information Center, National Institute of Health and Nutrition, Tokyo, Japan
| | - Koichi Kokame
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Toshiyuki Miyata
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Eiji Kaneko
- Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Kentaro Shimokado
- Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
- * E-mail:
| |
Collapse
|
17
|
Shimo T, Adachi Y, Yamanouchi S, Tsuji S, Kimata T, Umezawa K, Okigaki M, Takaya J, Ikehara S, Kaneko K. A novel nuclear factor κB inhibitor, dehydroxymethylepoxyquinomicin, ameliorates puromycin aminonucleoside-induced nephrosis in mice. Am J Nephrol 2013; 37:302-309. [PMID: 23548793 DOI: 10.1159/000348803] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 02/08/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Minimal-change nephrotic syndrome (MCNS) is a kidney disease defined by selective proteinuria and hypoalbuminemia occurring in the absence of cellular glomerular infiltrates or immunoglobulin deposits. Recent observations suggest that nuclear factor κB (NF-κB) of podocyte is strongly associated with the development of proteinuria in MCNS. Dehydroxymethylepoxyquinomicin (DHMEQ) is a novel NF-κB inhibitor that potently inhibits DNA-binding activity of NF-κB, resulting in several therapeutic effects in various pathological conditions. We conducted this study to ask whether DHMEQ may ameliorate the nephrosis in mice induced by puromycin aminonucleoside (PAN), which is considered to be an animal model for MCNS. METHODS/RESULTS Pretreatment with DHMEQ alleviated the proteinuria and reversed the serum abnormalities in mice nephrosis induced by 450 mg/kg of PAN. Increased serum interleukin-6 level in PAN-induced nephrosis was also completely suppressed by DHMEQ. Electron microscopic analyses of glo-meruli indicated that DHMEQ can inhibit the podocyte foot process effacement via blocking the translocation of podocyte NF-κB from cytoplasm to nucleus. CONCLUSIONS These results suggest that DHMEQ can be a potential therapeutic agent for MCNS.
Collapse
Affiliation(s)
- Tomohiko Shimo
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Shimizu K, Konno S, Ozaki M, Umezawa K, Yamashita K, Todo S, Nishimura M. Dehydroxymethylepoxyquinomicin (DHMEQ), a novel NF-kappaB inhibitor, inhibits allergic inflammation and airway remodelling in murine models of asthma. Clin Exp Allergy 2012; 42:1273-81. [PMID: 22805475 DOI: 10.1111/j.1365-2222.2012.04007.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Dehydroxymethylepoxyquinomicin (DHMEQ) is a newly developed compound that inhibits nuclear factor κB activation and is reported to ameliorate animal models of various inflammatory diseases without significant adverse effects. Because nuclear factor κB is a transcription factor that plays a critical role in the pathophysiology of asthma, DHMEQ may be of therapeutic benefit in asthma. OBJECTIVE The purpose of this study was to evaluate the effects of DHMEQ on airway inflammation and remodelling in murine models of asthma. METHODS The BALB/c mice were sensitized and then challenged acutely or chronically with ovalbumin and administered DHMEQ intraperitoneally before each challenge. Inflammation of airways, lung histopathology and airway hyper responsiveness to methacholine challenge were evaluated. In addition, the effect of DHMEQ on production of cytokines and eotaxin-1 by murine splenocytes, human peripheral blood mononuclear cells and bronchial epithelial cells was investigated. RESULTS Airway hyper responsiveness was ameliorated in both acutely and chronically challenged models by treatment with DHMEQ. DHMEQ significantly reduced eosinophilic airway inflammation and levels of Th2 cytokines in bronchoalveolar lavage fluid in the acute model. It also inhibited parameters of airway remodelling including mucus production, peribronchial fibrosis and the expression of α-smooth muscle actin. Moreover, the production of Th2 cytokines from murine splenocytes and human peripheral blood mononuclear cells and the production of eotaxin-1 by bronchial epithelial cells were inhibited by DHMEQ. CONCLUSIONS AND CLINICAL RELEVANCE These results indicate that DHMEQ inhibits allergic airway inflammation and airway remodelling in murine models of asthma. DHMEQ may have therapeutic potential in the treatment of asthma.
Collapse
Affiliation(s)
- K Shimizu
- First Department of Medicine, Hokkaido University School of Medicine, Kita-ku, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
Vittone F, Liberman A, Vasic D, Ostertag R, Esser M, Walcher D, Ludwig A, Marx N, Burgmaier M. Sitagliptin reduces plaque macrophage content and stabilises arteriosclerotic lesions in Apoe (-/-) mice. Diabetologia 2012; 55:2267-75. [PMID: 22648661 DOI: 10.1007/s00125-012-2582-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 04/16/2012] [Indexed: 02/04/2023]
Abstract
AIMS/HYPOTHESIS Inhibitors of dipeptidyl peptidase-IV (DPP-IV), such as sitagliptin, increase glucagon-like peptide-1 (GLP-1) concentrations and are current treatment options for patients with type 2 diabetes mellitus. As patients with diabetes exhibit a high risk of developing severe atherosclerosis, we investigated the effect of sitagliptin on atherogenesis in Apoe (-/-) mice. METHODS Apoe (-/-) mice were fed a high-fat diet and treated with either sitagliptin or placebo for 12 weeks. Plaque size and plaque composition were analysed using Oil Red O staining and immunohistochemistry. Furthermore, in vitro experiments with the modified Boyden chamber and with gelatine zymography were performed to analyse the effects of GLP-1 on isolated human monocyte migration and metalloproteinase-9 (MMP-9) release. RESULTS Treatment of Apoe (-/-) mice with sitagliptin significantly reduced plaque macrophage infiltration (the aortic root and aortic arch both showing a 67% decrease; p < 0.05) and plaque MMP-9 levels (aortic root showing a 69% and aortic arch a 58% reduction; both p < 0.01) compared with controls. Moreover, sitagliptin significantly increased plaque collagen content more than twofold (aortic root showing an increase of 58% and aortic arch an increase of 73%; both p < 0.05) compared with controls but did not change overall lesion size (8.1 ± 3.5% vs 5.1 ± 2.5% for sitagliptin vs controls; p=NS). In vitro, pretreatment of isolated human monocytes with GLP-1 significantly decreased cell migration induced by both monocyte chemotactic protein-1 and by the protein known as regulated on activation, normal T cell expressed and secreted (RANTES) in a concentration-dependent manner. Furthermore, GLP-1 significantly decreased MMP-9 release from isolated human monocyte-derived macrophages. CONCLUSIONS/INTERPRETATION Sitagliptin reduces plaque inflammation and increases plaque stability, potentially by GLP-1-mediated inhibition of chemokine-induced monocyte migration and macrophage MMP-9 release. The effects observed may provide potential mechanisms for how DPP-IV inhibitors could modulate vascular disease in high-risk patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- F Vittone
- Department of Internal Medicine II - Cardiology, University of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Schleithoff C, Voelter-Mahlknecht S, Dahmke IN, Mahlknecht U. On the epigenetics of vascular regulation and disease. Clin Epigenetics 2012; 4:7. [PMID: 22621747 PMCID: PMC3438017 DOI: 10.1186/1868-7083-4-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 03/09/2012] [Indexed: 12/16/2022] Open
Abstract
Consolidated knowledge is accumulating as to the role of epigenetic regulatory mechanisms in the physiology of vascular development and vascular tone as well as in the pathogenesis of cardiovascular disease. The modulation of gene expression through modification of the epigenome by structural changes of the chromatin architecture without alterations of the associated genomic DNA sequence is part of the cellular response to environmental changes. Such environmental conditions, which are finally being translated into adaptations of the cardiovascular system, also comprise pathological conditions such as atherosclerosis or myocardial infarction. This review summarizes recent findings on the epigenetics of vascular regulation and disease and presents nutritional and pharmacological approaches as novel epigenetic strategies in the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Christina Schleithoff
- Saarland University Medical Center, Department of Internal Medicine, Division of Immunotherapy and Gene Therapy, Homburg, Saar, D-66421, Germany
| | - Susanne Voelter-Mahlknecht
- Institute of Occupational and Social Medicine and Health Services Research, University of Tuebingen, Wilhelmstrasse 27, D-72074, Tuebingen, Germany
| | - Indra Navina Dahmke
- Saarland University Medical Center, Department of Internal Medicine, Division of Immunotherapy and Gene Therapy, Homburg, Saar, D-66421, Germany
| | - Ulrich Mahlknecht
- Saarland University Medical Center, Department of Internal Medicine, Division of Immunotherapy and Gene Therapy, Homburg, Saar, D-66421, Germany
| |
Collapse
|
21
|
Ohno O, Suenaga K, Uemura D. Secondary metabolites with new medicinal functions from marine organisms. ADVANCES IN FOOD AND NUTRITION RESEARCH 2012; 65:185-93. [PMID: 22361187 DOI: 10.1016/b978-0-12-416003-3.00011-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
We have focused on the identification of natural key compounds that possess biologically and medicinally intriguing functions. Some of bioactive naturally occurring compounds isolated from marine organisms were found to possess unique biological activities. Halichlorine from the marine sponge Halichondria okadai was shown to inhibit the activity of nuclear factor-κB in endothelial cells and block L-type Ca²⁺ channels. Thus, it may have therapeutic potentials for diseases such as atherosclerosis and hypertension.
Collapse
Affiliation(s)
- Osamu Ohno
- Department of Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan.
| | | | | |
Collapse
|
22
|
Shimo T, Adachi Y, Umezawa K, Okigaki M, Takaya J, Taniuchi S, Ikehara S, Kaneko K. Dehydroxymethylepoxyquinomicin (DHMEQ) can suppress tumour necrosis factor-α production in lipopolysaccharide-injected mice, resulting in rescuing mice from death in vivo. Clin Exp Immunol 2011; 166:299-306. [PMID: 21985376 PMCID: PMC3219905 DOI: 10.1111/j.1365-2249.2011.04475.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2011] [Indexed: 12/01/2022] Open
Abstract
Dehydroxymethylepoxyquinomicin (DHMEQ), a new nuclear factor (NF)-κB inhibitor, has several beneficial effects, including the suppression of tumour growth and anti-inflammatory effects. DHMEQ can also suppress the production of tumour necrosis factor (TNF)-α induced by lipopolysaccharide (LPS) in vitro. In the present study, we examine the effects of DHMEQ on TNF-α production in vivo and on the survival of mice injected with LPS. When DHMEQ was injected into mice 2 h before LPS injection, the survival of the LPS-injected mice was prolonged. When DHMEQ was injected twice (2 h before LPS injection and the day after LPS injection), all the mice were rescued. The injection of DHMEQ 1 h after LPS injection and the day after LPS injection also resulted in the rescue of all mice. The serum levels of TNF-α in the mice that received both LPS and DHMEQ were suppressed compared to the mice that received only LPS. These results suggest that DHMEQ can be utilized for the prevention and treatment of endotoxin shock.
Collapse
Affiliation(s)
- T Shimo
- Department of Pediatrics, Kansai Medical University, Moriguchi City, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Wong MC, van Diepen JA, Hu L, Guigas B, de Boer HC, van Puijvelde GH, Kuiper J, van Zonneveld AJ, Shoelson SE, Voshol PJ, Romijn JA, Havekes LM, Tamsma JT, Rensen PCN, Hiemstra PS, Berbée JFP. Hepatocyte-specific IKKβ expression aggravates atherosclerosis development in APOE*3-Leiden mice. Atherosclerosis 2011; 220:362-8. [PMID: 21798539 DOI: 10.1016/j.atherosclerosis.2011.06.055] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 06/03/2011] [Accepted: 06/29/2011] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The liver is the key organ involved in systemic inflammation, but the relation between hepatic inflammation and atherogenesis is poorly understood. Since nuclear factor-κB (NF-κB) is a central regulator of inflammatory processes, we hypothesized that chronically enhanced hepatic NF-κB activation, through hepatocyte-specific expression of IκB kinase-β (IKKβ) (LIKK), will aggravate atherosclerosis development in APOE*3-Leiden (E3L) mice. METHODS AND RESULTS E3L.LIKK and E3L control littermates were fed a Western-type diet for 24 weeks. E3L.LIKK mice showed a 2.3-fold increased atherosclerotic lesion area and more advanced atherosclerosis in the aortic root with less segments without atherosclerotic lesions (11% vs. 42%), and more segments with mild (63% vs. 44%) and severe (26% vs. 14%) lesions. Expression of LIKK did not affect basal levels of inflammatory parameters, but plasma cytokine levels tended to be higher in E3L.LIKK mice after lipopolysaccharide (LPS) administration. E3L.LIKK mice showed transiently increased plasma cholesterol levels, confined to (V)LDL. This transient character resulted in a mild (+17%) increased cumulative plasma cholesterol exposure. CONCLUSION We conclude that selective activation of NF-κB in hepatocytes considerably promotes atherosclerosis development which is (at least partly) explained by an increased sensitivity to proinflammatory triggers and transiently increased plasma cholesterol levels.
Collapse
Affiliation(s)
- Man C Wong
- The Dept. of General Internal Medicine, Endocrinology, and Metabolic Diseases, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Direct Rel/NF-κB inhibitors: structural basis for mechanism of action. Future Med Chem 2011; 1:1683-707. [PMID: 21425986 DOI: 10.4155/fmc.09.96] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The Rel/NF-κB transcription factors have emerged as novel therapeutic targets for a variety of human diseases and pathological conditions, including inflammation, autoimmune diseases, cancer, ischemic injury, osteoporosis, transplant rejection and neurodegeneration. Several US FDA-approved drugs may, in part, attribute their therapeutic effects to the inhibition of the Rel/NF-κB pathway. Strategies for blocking the Rel/NF-κB signaling pathway have inspired the pharmaceutical industry to develop inhibitors for I-κB kinase, however, this article focuses instead on identifying natural compounds that directly target and inhibit DNA binding and transcription activity of Rel/NF-κB. These include compounds containing a quinone core, an α,β unsaturated carbonyl and a benzene diamine. By investigating the mechanisms of action of existing natural inhibitors, novel strategies and synthetic approaches can be devised that will facilitate the development of novel and selective Rel/NF-κB inhibitors with better safety profiles.
Collapse
|
25
|
Umezawa K. Possible role of peritoneal NF-κB in peripheral inflammation and cancer: Lessons from the inhibitor DHMEQ. Biomed Pharmacother 2011; 65:252-9. [DOI: 10.1016/j.biopha.2011.02.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 02/08/2011] [Indexed: 01/12/2023] Open
|
26
|
Gómez-Guerrero C, Mallavia B, Egido J. Targeting Inflammation in Cardiovascular Diseases. Still a Neglected field? Cardiovasc Ther 2011; 30:e189-97. [DOI: 10.1111/j.1755-5922.2011.00274.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
27
|
Imai T, Oikawa Y, Shimada A, Oguchi S, Takamiya Y, Katsuki T, Okubo Y, Osaki T, Tahara H, Matsushima Y, Miyazaki JI, Itoh H. Proatherogenic Effect of Interleukin-18 is Exerted with High-fat Diet, but not with Normal Diet in Spontaneously Hyperlipidemic Mice. J Atheroscler Thromb 2011; 18:1090-101. [DOI: 10.5551/jat.7567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
28
|
Kubota T, Fukuya Y, Hashimoto R, Kanda T, Suzuki H, Okamura Y, Nanki T, Miyasaka N, Umezawa K. Possible Involvement of Chemokine-induced Platelet Activation in Thrombophilic Diathesis of Antiphospholipid Syndrome. Ann N Y Acad Sci 2009; 1173:137-45. [DOI: 10.1111/j.1749-6632.2009.04648.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Etzion Y, Hackett A, Proctor BM, Ren J, Nolan B, Ellenberger T, Muslin AJ. An unbiased chemical biology screen identifies agents that modulate uptake of oxidized LDL by macrophages. Circ Res 2009; 105:148-57. [PMID: 19542016 DOI: 10.1161/circresaha.109.195818] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Macrophage-derived foam cells are thought to play a major role in atherosclerotic lesion formation and progression. An automated assay was established to evaluate the uptake of fluorescently labeled oxidized low-density lipoprotein (oxLDL) by a monocyte/macrophage cell line. The assay was used to screen 480 known bioactive compounds. Twenty-two active compounds were identified. Efficacy studies in peritoneal macrophages demonstrated a high rate of concordance with the initial screening results. Inhibitory compounds confirmed important previous findings and identified new drugs of interest including: 3 blockers of nuclear factor kappab activation, 2 protein kinase C inhibitors, a phospholipase C inhibitor, and 2 antipsychotic drugs. In addition, an opioid receptor agonist was found to increase the oxLDL uptake of macrophages. The involvement of nuclear factor kappaB in oxLDL uptake was validated in peritoneal macrophages in vivo. The results support a model in which oxLDL uptake is dependent on the activation of multiple intracellular signaling pathways that culminate in actin-mediated lipoprotein internalization.
Collapse
Affiliation(s)
- Yoram Etzion
- Center for Cardiovascular Research, Washington University School of Medicine, Box 8086, 660 South Euclid Ave, St Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Creus KK, De Paepe B, De Bleecker JL. Idiopathic inflammatory myopathies and the classical NF-κB complex: Current insights and implications for therapy. Autoimmun Rev 2009; 8:627-31. [DOI: 10.1016/j.autrev.2009.02.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 02/05/2009] [Indexed: 01/20/2023]
|
31
|
Mazière C, Mazière JC. Activation of transcription factors and gene expression by oxidized low-density lipoprotein. Free Radic Biol Med 2009; 46:127-37. [PMID: 18996472 DOI: 10.1016/j.freeradbiomed.2008.10.024] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/26/2008] [Accepted: 10/01/2008] [Indexed: 11/25/2022]
Abstract
It is well recognized that oxidized LDL (OxLDL) plays a crucial role in the initiation and progression of atherosclerosis. Many biological effects of OxLDL are mediated through signaling pathways, especially via the activation of transcription factors, which in turn stimulate the expression of genes involved in the inflammatory and oxidative stress response or in cell cycle regulation. In this review, we will discuss the various transcription factors activated by OxLDL, the studied cell types, the active compounds of the OxLDL particle, and the downstream genes when identified. Identification of the transcription factors and some of the downstream genes regulated by OxLDL has helped us understand the molecular mechanism involved in generation of the atherosclerotic plaque.
Collapse
Affiliation(s)
- Cécile Mazière
- Biochemistry Laboratory, North Hospital, University of Picardie-Jules Verne, and INSERM, ERI 12, Amiens F-80000, France.
| | | |
Collapse
|
32
|
Cheng SM, Lai JH, Yang SP, Tsao TP, Ho LJ, Liou JT, Cheng CC. Modulation of human T cells signaling transduction by lovastatin. Int J Cardiol 2008; 140:24-33. [PMID: 19046782 DOI: 10.1016/j.ijcard.2008.10.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 07/23/2008] [Accepted: 10/26/2008] [Indexed: 11/26/2022]
Abstract
Statins are applied clinically to treat hypercholesterolemia and proposed to have some kinds of anti-inflammatory properties for reducing the incidence of atherosclerosis-related cardiovascular events. However, it was rarely known about statins on the signal transduction on human primary T cells. To gain insight into the mechanism of statins on human T cells, we investigated the effects of both lovastatin and atorvastatin on activated human primary T cells. The human primary T cells from the blood of normal human beings were isolated. We found that lovastatin, but not atorvastatin, can dose-dependently inhibit cytokine production such as interleukin-2, interleukin-4, and interferon-gamma from activated human T cells. Neither lovastatin nor atorvastatin can regulate the TNF-alpha production on both activated human T cells and monocytes. Molecular investigation was performed that lovastatin, but not atorvastatin, could down-regulate both activator protein-1 and NF-kappaB DNA binding activities, assessed by electrophoretic mobility shift assay. Our observations may extend potential and differential therapeutic mechanisms of lovastatin with cell-mediated capacity to prevent or treat some of inflammation related diseases.
Collapse
Affiliation(s)
- Shu-Meng Cheng
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center No 325, Section 2, Cheng-Kung Road, Neihu 114, Taipei, Taiwan, ROC.
| | | | | | | | | | | | | |
Collapse
|
33
|
Gareus R, Kotsaki E, Xanthoulea S, van der Made I, Gijbels MJJ, Kardakaris R, Polykratis A, Kollias G, de Winther MPJ, Pasparakis M. Endothelial cell-specific NF-kappaB inhibition protects mice from atherosclerosis. Cell Metab 2008; 8:372-83. [PMID: 19046569 DOI: 10.1016/j.cmet.2008.08.016] [Citation(s) in RCA: 315] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 07/09/2008] [Accepted: 08/21/2008] [Indexed: 10/21/2022]
Abstract
Atherosclerosis is a progressive disorder of the arterial wall and the underlying cause of cardiovascular diseases such as heart attack and stroke. Today, atherosclerosis is recognized as a complex disease with a strong inflammatory component. The nuclear factor-kappaB (NF-kappaB) signaling pathway regulates inflammatory responses and has been implicated in atherosclerosis. Here, we addressed the function of NF-kappaB signaling in vascular endothelial cells in the pathogenesis of atherosclerosis in vivo. Endothelium-restricted inhibition of NF-kappaB activation, achieved by ablation of NEMO/IKKgamma or expression of dominant-negative IkappaBalpha specifically in endothelial cells, resulted in strongly reduced atherosclerotic plaque formation in ApoE(-/-) mice fed with a cholesterol-rich diet. Inhibition of NF-kappaB abrogated adhesion molecule induction in endothelial cells, impaired macrophage recruitment to atherosclerotic plaques, and reduced expression of cytokines and chemokines in the aorta. Thus, endothelial NF-kappaB signaling orchestrates proinflammatory gene expression at the arterial wall and promotes the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Ralph Gareus
- Institute of Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47, 50674 Cologne, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hayashi K. [Kidney disease: potential of anti-inflammatory approaches for drug therapy]. Nihon Yakurigaku Zasshi 2008; 132:89-95. [PMID: 18689957 DOI: 10.1254/fpj.132.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
35
|
Kubota T, Hoshino M, Aoki K, Ohya K, Komano Y, Nanki T, Miyasaka N, Umezawa K. NF-kappaB inhibitor dehydroxymethylepoxyquinomicin suppresses osteoclastogenesis and expression of NFATc1 in mouse arthritis without affecting expression of RANKL, osteoprotegerin or macrophage colony-stimulating factor. Arthritis Res Ther 2008; 9:R97. [PMID: 17892600 PMCID: PMC2212584 DOI: 10.1186/ar2298] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 08/25/2007] [Accepted: 09/25/2007] [Indexed: 12/31/2022] Open
Abstract
Inhibition of NF-κB is known to be effective in reducing both inflammation and bone destruction in animal models of arthritis. Our previous study demonstrated that a small cell-permeable NF-κB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), suppresses expression of proinflammatory cytokines and ameliorates mouse arthritis. It remained unclear, however, whether DHMEQ directly affects osteoclast precursor cells to suppress their differentiation to mature osteoclasts in vivo. The effect of DHMEQ on human osteoclastogenesis also remained elusive. In the present study, we therefore examined the effect of DHMEQ on osteoclastogenesis using a mouse collagen-induced arthritis model, and using culture systems of fibroblast-like synovial cells obtained from patients with rheumatoid arthritis, and of osteoclast precursor cells from peripheral blood of healthy volunteers. DHMEQ significantly suppressed formation of osteoclasts in arthritic joints, and also suppressed expression of NFATc1 along the inner surfaces of bone lacunae and the eroded bone surface, while serum levels of soluble receptor activator of NF-κB ligand (RANKL), osteoprotegerin and macrophage colony-stimulating factor were not affected by the treatment. DHMEQ also did not suppress spontaneous expression of RANKL nor of macrophage colony-stimulating factor in culture of fibroblast-like synovial cells obtained from patients with rheumatoid arthritis. These results suggest that DHMEQ suppresses osteoclastogenesis in vivo, through downregulation of NFATc1 expression, without significantly affecting expression of upstream molecules of the RANKL/receptor activator of NF-κB/osteoprotegerin cascade, at least in our experimental condition. Furthermore, in the presence of RANKL and macrophage colony-stimulating factor, differentiation and activation of human osteoclasts were also suppressed by DHMEQ, suggesting the possibility of future application of NF-κB inhibitors to rheumatoid arthritis therapy.
Collapse
Affiliation(s)
- Tetsuo Kubota
- Department of Microbiology and Immunology, Tokyo Medical and Dental University Graduate School of Health Sciences, Tokyo, Japan
| | - Machiko Hoshino
- Department of Microbiology and Immunology, Tokyo Medical and Dental University Graduate School of Health Sciences, Tokyo, Japan
| | - Kazuhiro Aoki
- Department of Hard Tissue Engineering, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Keiichi Ohya
- Department of Hard Tissue Engineering, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Yukiko Komano
- Department of Medicine and Rheumatology, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Toshihiro Nanki
- Department of Medicine and Rheumatology, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Nobuyuki Miyasaka
- Department of Medicine and Rheumatology, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Kazuo Umezawa
- Department of Applied Chemistry, Keio University, Kanagawa, Japan
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Inflammation contributes to the formation and progression of atherosclerosis and the therapeutic potential of some anti-inflammatory drugs has been evaluated for possible antiatherosclerotic effects. This review will briefly describe the mechanisms underlying the inflammation-atherosclerosis connection, the effect of various anti-inflammatory therapies on atherosclerotic disease and a sampling of the potential targets and agents under evaluation. RECENT FINDINGS Some agents with anti-inflammatory properties appear to have beneficial effects on atherosclerosis or subsequent risk for cardiovascular events, while others have been disappointing. The anti-inflammatory actions of statins have been linked retrospectively with their favorable effects on atherosclerosis progression and clinical outcomes. The cardiovascular safety of COX-2 inhibitors is being assessed prospectively in patients with atherosclerosis. Potential new therapeutic agents targeting other inflammatory mechanisms and oxidative stress are being evaluated in animal models and clinical trials. SUMMARY Due to the contributory inflammatory pathways in atherosclerosis, the properties of existing and novel anti-inflammatory agents are being carefully and actively evaluated in cardiovascular disease. Advances in our understanding of both atherosclerosis and the inflammatory contributors may play an important role in future strategies to decrease the incidence of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Sami P Moubayed
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada
| | | | | |
Collapse
|